integrated molecular characterization of fumarate hydratase … · 2021. 1. 7. · 6 1. of. 429 rcc...

34
1 Integrated Molecular Characterization of Fumarate Hydratase-deficient Renal Cell Carcinoma Guangxi Sun 1,# , Xingming Zhang 1,# , Jiayu Liang 1,# , Xiuyi Pan 2,# , Sha Zhu 1,# , Zhenhua Liu 1,# , Cameron M. Armstrong 3 , Jianhui Chen 4 , Wei Lin 5 , Banghua Liao 1 , Tianhai Lin 1 , Rui Huang 6 , Mengni Zhang 2 , Linmao Zheng 2 , Xiaoxue Yin 2 , Ling Nie 2 , Pengfei Shen 1 , Jinge Zhao 1 , Haoran Zhang 1 , Jindong Dai 1 , Yali Shen 7 , Zhiping Li 7 , Jiyan Liu 8 , Junru Chen 1 , Jiandong Liu 1,9 , Zhipeng Wang 1 , Xudong Zhu 1 , Yuchao Ni 1 , Dan Qin 10 , Ling Yang 11 , Yuntian Chen 11 , Qiang Wei 1 , Xiang Li 1 , Qiao Zhou 2 , Haojie Huang 12,, Jin Yao 11,, *, Ni Chen 2,, *, Hao Zeng 1,, * 1. Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China 2. Department of Pathology, West China Hospital, Sichuan University, 610041, Chengdu, China 3. Department of Urology and Comprehensive Cancer Center, University of California Davis, Sacramento, 95817, CA, USA 4. Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China 5. Department of Urology, Zigong Fourth People’s Hospital, 643000, Zigong, China 6. Department of Nuclear medicine, West China Hospital, Sichuan University, 610041, Chengdu, China 7. Department of Oncology, West China Hospital, Sichuan University, 610041, Chengdu, China 8. Department of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China Research. on July 3, 2021. © 2021 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

Upload: others

Post on 12-Feb-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Integrated Molecular Characterization of Fumarate Hydratase-deficient

    Renal Cell Carcinoma

    Guangxi Sun 1,#, Xingming Zhang 1,#, Jiayu Liang 1,#, Xiuyi Pan 2,#, Sha Zhu 1,#,

    Zhenhua Liu 1,#, Cameron M. Armstrong 3, Jianhui Chen 4, Wei Lin 5, Banghua

    Liao 1, Tianhai Lin 1, Rui Huang 6, Mengni Zhang 2, Linmao Zheng 2, Xiaoxue

    Yin 2, Ling Nie 2, Pengfei Shen 1, Jinge Zhao 1, Haoran Zhang 1, Jindong Dai 1,

    Yali Shen 7, Zhiping Li 7, Jiyan Liu 8, Junru Chen 1, Jiandong Liu 1,9, Zhipeng

    Wang 1, Xudong Zhu 1, Yuchao Ni 1, Dan Qin 10, Ling Yang 11, Yuntian Chen 11,

    Qiang Wei 1, Xiang Li 1, Qiao Zhou 2, Haojie Huang 12,+, Jin Yao 11,

    +,*, Ni Chen

    2,+,*, Hao Zeng 1,+,*

    1. Department of Urology, Institute of Urology, West China Hospital, Sichuan

    University, 610041, Chengdu, China

    2. Department of Pathology, West China Hospital, Sichuan University, 610041,

    Chengdu, China

    3. Department of Urology and Comprehensive Cancer Center, University of

    California Davis, Sacramento, 95817, CA, USA

    4. Department of Urology, Fujian Medical University Union Hospital, Fuzhou,

    China

    5. Department of Urology, Zigong Fourth People’s Hospital, 643000, Zigong,

    China

    6. Department of Nuclear medicine, West China Hospital, Sichuan University,

    610041, Chengdu, China

    7. Department of Oncology, West China Hospital, Sichuan University, 610041,

    Chengdu, China

    8. Department of Biotherapy, West China Hospital, Sichuan University,

    610041, Chengdu, China

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 2

    9. Department of Urology, The First Affiliated Hospital, School of Medicine,

    Zhejiang University, 310009, Hangzhou, China

    10. The Bioinformatics Department, Basebiotech Co., Ltd, 610000, Chengdu,

    China

    11. Department of Radiology, West China Hospital, Sichuan University, 610041,

    Chengdu, China

    12. Departments of Biochemistry and Molecular Biology and Urology, Mayo

    Clinic College of Medicine and Science, Rochester, 55905, MN, USA

    # These authors are co-first authors and contributed equally.

    + These authors are co-senior authors.

    * These authors are co-corresponding authors.

    Running title

    Genomic and epigenomic profiling of FH-deficient RCC

    Corresponding Author:

    Address: No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan

    Province, PR China; Phone number: +86 28 85422026; Fax: +86 28

    85582944.

    Email address: [email protected] (Jin. Yao); [email protected] (Ni. Chen);

    [email protected] (Hao. Zeng)

    Conflicts of interest

    The authors declare no conflicts of interest.

    Words count: 3,903 words

    Total number of Figures and Tables: 4 figures and 1 table in main

    manuscript, 6 figures and 4 tables in supplementary appendix.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    mailto:[email protected]:[email protected]:[email protected]://clincancerres.aacrjournals.org/

  • 3

    Translational Relevance

    Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare

    but lethal malignancy, and its genetic basis is poorly understood. We

    comprehensively profiled untreated primary FH-deficient RCC tissues to

    characterize the molecular landscape. We identified that DNA methylation

    profiles could be the biological foundation for tumor pathogenesis; hence,

    compared with other RCC subtypes, methylation sequencing should be

    recommended for patients with FH-deficient RCC. Moreover, FH-deficient

    RCC was characterized by an immunogenic phenotype despite its low overall

    mutation burden. Patients with this tumor type could benefit from immune

    checkpoint blockade-based strategies. Our results provide insights into the

    biology of FH-deficient RCC and pave the way for the development of novel

    promising therapies.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 4

    Abstract

    Purpose: Fumarate hydratase-deficient renal cell carcinoma (FH-deficient

    RCC) is a rare but lethal subtype of RCC. Little is known about the genomic

    profile of FH-deficient RCC, and the therapeutic options for advanced disease

    are limited. To this end, we performed a comprehensive genomics study to

    characterize the genomic and epigenomic features of FH-deficient RCC.

    Experimental Design: Integrated genomic, epigenomic, and molecular

    analyses were performed on 25 untreated primary FH-deficient RCCs.

    Complete clinicopathological and follow-up data of these patients were

    recorded.

    Results: We identified that FH-deficient RCC manifested low somatic mutation

    burden (median 0.58 mutations per megabase), but with frequent somatic

    copy number alterations. The majority of FH-deficient RCCs were

    characterized by a CpG sites island methylator phenotype (CIMP), displaying

    concerted hypermethylation at numerous CpG sites in genes of transcription

    factors, tumor-suppressors and tumor hallmark pathways. However, a few

    cases (20%) with low metastatic potential showed relatively low DNA

    methylation levels, indicating the heterogeneity of methylation pattern in

    FH-deficient RCC. Moreover, FH-deficient RCC is potentially highly

    immunogenic, characterized by increased tumor T cell infiltration but high

    expression of immune checkpoint molecules in tumors. Clinical data further

    demonstrated that patients receiving immune checkpoint blockade-based

    treatment achieved improved progression free-survival over those treated with

    antiangiogenic monotherapy (median: 13.3 vs. 5.1 months, P = 0.03).

    Conclusions: These results reveal the genomic features and provide new

    insight into potential therapeutic strategies for FH-deficient RCC.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 5

    Introduction 1

    Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is 2

    a rare subtype of kidney cancer characterized by pathologic germline/somatic 3

    mutation of the fumarate hydratase (FH) gene, with a lack of FH staining on 4

    immunohistochemistry (IHC). FH-deficient RCC is difficult to diagnose, since it 5

    frequently shows mixed pathologically architectural patterns; therefore, it is 6

    highly susceptible to be misdiagnosed as other RCC subtypes (1,2). Clinically, 7

    most cases of FH-deficient RCC are very aggressive and frequently present 8

    with locally advanced or metastatic disease at initial diagnosis (3). 9

    Unfortunately, owing to its rarity, little is known about its genetic basis, and 10

    there are currently no standard treatment strategies for patients with advanced 11

    disease. 12

    To date, our knowledge of the genetic basis of FH-deficient RCC mainly 13

    comes from the reports of the Cancer Genome Atlas (TCGA) database. Using 14

    comprehensive genomic analyses, TCGA identified a distinct group of papillary 15

    RCC characterized by a CpG island methylator phenotype (CIMP), of which 16

    five cases were found to harbor germline or somatic FH mutation (4). However, 17

    due to the limited cases in the TCGA cohort, specific research aiming to 18

    explore the genomic landscape of FH-deficient RCC is still lacking. In this 19

    study, we performed a high-throughput genomic and epigenomic analysis of 20

    untreated primary FH-deficient RCC tumors from multi-institutional patient 21

    cohorts in order to provide molecular insights into FH-deficient RCC and 22

    suggest paths for the development of biology-driven precision medicine. 23

    24

    Materials and Methods 25

    Patient Identification 26

    We cooperated with nine medical centers to collect FH-deficient RCC 27

    samples. All suspicious RCC tumors were referred for pathologic consultation 28

    at our medical center (West China Hospital). Between 2014 and 2019, a total 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 6

    of 429 RCC samples were retrospectively analyzed (Figure S1). All candidate 1

    cases were reviewed by two experienced uropathologists (Ni Chen and 2

    Mengni Zhang) and subsequently subjected to immunohistochemical staining 3

    (IHC) of FH protein. Diagnosis of FH-deficient RCC was confirmed by Sanger 4

    sequencing for FH mutation (Figure S2). Clinicopathological data were 5

    retrospectively collected. A total of 25 cases with FH-deficient RCC were 6

    identified. Untreated primary tumor tissues and matched germline samples 7

    were collected. DNA and methylation sequencing of formalin-fixed 8

    paraffin-embedded (FFPE) tissues were subsequently performed. Twenty-two 9

    FFPE tumor tissues and matched adjacent normal tissues (n = 12) or blood 10

    samples (n = 10) were collected for whole-exome sequencing (WES) to 11

    assess the genomic mutation characteristics. Targeted sequencing was 12

    performed on two biopsy tumor tissues (FHRCC18 and FHRCC24) and 13

    matched blood samples due to insufficient tumor DNA for WES. One patient 14

    (FHRCC14) had only a blood sample for WES due to insufficient tumor tissue. 15

    EPIC array was performed on 20 tumors and 12 adjacent normal tissues. The 16

    study was conducted in accordance with the Declaration of Helsinki and all 17

    clinical samples were acquired with written informed consents under 18

    permission from the Ethics Committee of West China Hospital of Sichuan 19

    University. All patients or family members provided written consent for genetic 20

    analysis. 21

    22

    Clinicopathological characteristics 23

    Clinicopathological data were retrospectively collected, including age, 24

    gender, family history, metastatic sites, TNM stage, histological type, ISUP 25

    grade, surgery type, and systemic treatment type. Synchronous metastasis 26

    was defined as the diagnosis of a distant metastasis at the time of the primary 27

    renal cell carcinoma diagnosis. Metachronous metastasis was defined as the 28

    metastasis occurred after three months postoperatively. 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 7

    1

    Outcomes 2

    Patients visited the urologists for regular evaluations (physical 3

    examination, radiologic assessment and laboratory tests) every 4-6 weeks. 4

    Response was defined by Response Evaluation Criteria in Solid Tumors 5

    (RECIST) version 1.1 (5). The end-points were objective response rate (ORR), 6

    disease control rate (DCR), disease-free survival (DFS), progression-free 7

    survival (PFS), and overall survival (OS). ORR and DCR were assessed 8

    according to RECIST criteria 1.1. ORR was defined as the proportion of 9

    patients with complete or partial responses as the best radiological response 10

    to therapy. DCR encompassed the proportion of patients who achieved an 11

    objective response plus stable disease. DFS was defined as the time from 12

    diagnosis to local or regional recurrence or distance metastasis or death from 13

    any cause. PFS was defined as the time from initial diagnosis to disease 14

    progression or death. First-line PFS was defined as the time from the start of 15

    first-line systemic treatment to disease progression or death. OS was defined 16

    as the time from initial diagnosis to all-cause death. Systemic treatment OS 17

    was defined as the time from the start of systemic treatment to all-cause death. 18

    19

    WES and methylation sequencing 20

    For samples performing WES, high-quality genomic DNA was extracted 21

    using the GeneRead DNA FFPE Kit (180134, QIAGEN, Hilden, GER) 22

    according to the manufacturer’s specifications. Exome capture was performed 23

    using Agilent SureSelect Human All ExonV6 kit (Technologies, CA, USA) 24

    followed by paired-end sequencing using the Illumina Hiseq Xten platform 25

    (Illumine Inc, CA, USA). For panel sequencing, DNA was extracted and 26

    estimated by a targeted sequencing strategy capturing all exons of 642 27

    tumor-related genes (Table S1). Human Infinium MethylationEPIC BeadChip 28

    (Illumina) was used to analyze DNA methylation. Extracted DNA was treated 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 8

    with RNase A/T1 Mix (Thermo Scientific, Oberhausen, Germany) and 1

    subsequently purified using the Genomic DNA Clean & Concentrator™−10 Kit 2

    (Zymo Research, CA, USA). Bisulfite pyrosequencing was used to validate the 3

    results of the microarray analysis. The WES data generated in this paper have 4

    been deposited at the NCBI Sequence Read Archive (SRA) hosted by the NIH 5

    (SRA accession: PRJNA533711). The methylation data reported in this paper 6

    were deposited in the Gene Expression Omnibus (GEO) database (accession 7

    numbers GSE155207). More detailed materials and methods can be found in 8

    the supplementary methods. 9

    10

    Immunohistochemistry (IHC) 11

    IHC for FH, CD8, CD4 and PD-L1 were performed using an automatic 12

    staining platform, Ventana NexES (Roche, Basel, CH). Commercially available 13

    primary anti-FH (1: 800, sc-100743, Santa Cruz, TX, USA), CD8 (1:100, clone 14

    C8/144B, Dako, Copenhagen, DEN), CD4 (ZM-0418, 1:100, Zhongshan, 15

    Beijing, CHN) and PD-L1 (740-4859, 1:40, Roche) were used in this study. 16

    PD-L1 was determined by quantification of the density of positive cells (defined 17

    as the number of positive tumor cells per mm2) and the percentage of tumor 18

    cells. PD-L1 positivity was defined as PD-L1 expression ≥ 1%. CD4 and CD8 19

    were determined by quantification of the density of positive cells defined as the 20

    number of positive tumor infiltrating lymphocytes (TIL) per mm2 and the 21

    percentage of TILs. 22

    23

    Multiple immunofluorescence 24

    Multiplex staining and multispectral imaging were performed to identify the 25

    different types of immune cells. Multiplex immunofluorescence staining was 26

    obtained using PANO 7-plex kit (0004100100, Panovue, Beijing, CHN). 27

    Different primary antibodies, including CD8 (clone C8/144B, Panovue), CD56 28

    (clone 123C3, Panovue), HLA-DR (clone EPR3692, Panovue,), CD68 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 9

    (Panovue, clone BP6036) and PanCK (0057000050, Panovue) were 1

    sequentially applied, followed by horseradish peroxidase-conjugated 2

    secondary antibody incubation and tyramide signal amplification. The slides 3

    were microwave heat-treated after each Trichostatin A (TSA) operation. Nuclei 4

    were stained with 4′-6′-diamidino-2-phenylindole (D9542, DAPI, Sigma, 5

    Darmstadt, GER) after all the human antigens had been labeled. The stained 6

    slides were scanned via the Mantra System to obtain multispectral images 7

    (PerkinElmer, Massachusetts, USA). The scans were combined to build a 8

    single stack image. All slides were scanned at an absolute magnification of × 9

    200 and × 100. Images of unstained and single-stained sections were used to 10

    extract the spectrum of autofluorescence of tissues and each fluorescein, 11

    respectively. The extracted images were further used to establish a spectral 12

    library required for multispectral unmixing by Inform image analysis software 13

    (PerkinElmer). The CD8, macrophage and NK cells expression were quantified 14

    by the number of positive cells per mm2 and by the number of positive cells per 15

    number of nucleated cells. 16

    17

    Sanger sequencing 18

    The presence of somatic mutations of FH was validated by Sanger 19

    sequencing. For this analysis, primers to amplify a 220-bp fragment covering 20

    each exon (1-10) of FH gene were designed (Table S2). PCR was performed 21

    with the following thermal cycling conditions: a 95°C for 5 min; 35 cycles of 22

    95°C for 20 s; 60°C for 30 s and 72°C for 30 s; and a 72°C for 7 min. PCR 23

    amplification of 10ng of genomic DNA was performed using the Premix Taq™ 24

    (RR901A, Takara, Shiga, JPN) on a Veriti96 PCR Cycler (Life Technologies, 25

    Massachusetts, USA). PCR fragments were purified with ChargeSwitch™ 26

    PCR Clean-Up Kit (CS12000, Invitrogen, Oberhausen, GER), and the 27

    sequencing reactions were performed on an ABI 3730XL automatic sequencer 28

    (Life Technologies) according to the manufacturer’s instructions. All analyses 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 10

    were performed in duplicate. Sequences of the forward and reverse strands 1

    were analyzed using Chromas (Technelysium, AUS). 2

    3

    Bisulfite pyrosequencing 4

    Bisulfite modification of genomic DNA isolated from 30 samples was 5

    performed by standard methods. PCR primers were designed with the 6

    PyroMark Assay Design V.2.0 software (QIAGEN) (Table S3). PCR products 7

    were pyrosequenced with the PyromarkTM Q24 system (QIAGEN), according 8

    to the manufacturer’s protocol. 9

    10

    Statistics 11

    Fisher’s exact test was used to compare the difference between 12

    categorical variables. Median survival was estimated using the Kaplan–Meier 13

    method and the difference was tested using the log-rank test. All tests were 14

    two-sided and performed using R software v.3.2.3, SPSS v.16.0 and 15

    GraphPad V.6.0. A P-value < 0.05 was considered statistically significant. 16

    17

    Results 18

    Patient characteristics 19

    From 2014 to 2019, a total of 25 patients with FH-deficient RCC were 20

    identified from 429 suspicious cases (Figure S1A). Among them, 16 (64%) 21

    patients had germline FH mutations, eight (32%) patients had somatic FH 22

    mutations, and one patient (FHRCC20) had a germline mutation (c.1256C>T) 23

    and a second hit of an additional frameshift mutation (c.642delA) in the tumor. 24

    Baseline characteristics of all patients are summarized in Table 1 and S4. The 25

    median age at initial diagnosis was 38 years (range: 13-66 years) and the male: 26

    female ratio was 1.8:1. Male patients tended to be older at onset of primary 27

    RCC (P = 0.026, Figure S1B). At the end of follow-up (median: 14.3 months, 28

    range 3.8-137.1 months), 21 patients (84%, 21/25) presented with metastases, 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 11

    including 14 synchronous metastases and 7 metachronous metastases. The 1

    most common metastases were lymph nodes (76%, 16/21) and bones (48%, 2

    10/21). Five patients died by the end of follow-up. We found that those 3

    diagnosed over the age of 40 years old had a higher incidence of metastasis 4

    (P = 0.032, Figure S1C) and worse prognosis compared with the younger 5

    patients (Figure S1D and E). 6

    7

    The somatic mutational landscape of FH-deficient RCC 8

    WES was performed on 22 untreated primary tumors and matched 9

    germline samples. A median of 31 somatic mutations per tumor (range, 1-156) 10

    was identified, corresponding to an average of 0.58 mutations per megabase 11

    (range, 0.02–2.60) (Figure 1A). A total of 27 FH mutations were identified, 12

    including 15 missense mutations, 5 frame-shift indels, 5 nonsense mutations, 13

    1 splice site change, and 1 large deletion (Figure 1B). Apart from FH, the most 14

    frequently mutated genes in FH-deficient RCC were TTN (17%, 4/24) and NF2 15

    (13%, 3/24). Assessment of mutations within pathways demonstrated a high 16

    number of alterations involving chromatin modification (33%, 8/24), 17

    RTK-RAS-PI3K (33%, 8/24) and DNA damage repair pathway (29%, 7/24). 18

    Signature analysis identified that signature 6 contributed the most to these 19

    base substitutions, followed by signature 22 and signature 7, indicating that the 20

    defective DNA mismatch repair (signature 6), ultraviolet light exposure 21

    (signature 7) and aristolochic acid exposure (signature 22) might contribute to 22

    in the development of FH-deficient RCC (Figure S3A and B). 23

    Somatic copy number alterations (SCNAs) analysis revealed that 24

    FH-deficient RCC had frequent copy number loss of 1p, 8q, 10p, 15q and gain 25

    of 4p, 7q, 11q, and 19p (Figure 1C). Of note, frequent deletion of genes 26

    involving chromatin modification (KDM4B and KDM5A), SWI/SNF complex 27

    (SMARCA4 and ARID1A), DNA demethylases (TET1) and DNA damage repair 28

    pathway (ERCC1, ERCC2, XRCC1 and ATM) and cell cycle regulation 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 12

    (CDKN2C and CDKN2D) were identified in this cohort (Figure 1C). 1

    We further investigated the relationship between somatic mutations, 2

    SCNAs, and clinical features. Compared with young patients (< 40 years old), 3

    tumors of older patients (≥ 40 years old) tended to have a higher somatic 4

    mutation burden (median mutations per megabase: 0.75 vs. 0.27, Figure 1A); 5

    however, the difference did not reach a statistical significance (P = 0.051). 6

    Patients older than 40 years old also had increased chromatin instability than 7

    the younger patients (P = 0.047, Figure S3C and D). These results may reveal 8

    why older patients have poorer outcomes. 9

    10

    DNA methylation alterations associated with the pathogenesis of 11

    FH-deficient RCC 12

    EPIC array was performed on 20 FH-deficient RCC tumors with adjacent 13

    normal tissues. The study of the methylation profiles revealed that FH-deficient 14

    RCCs had a sharp global hypermethylation compared to adjacent normal 15

    tissues in all epigenomic structures, except for open sea (Figure S4A). In 16

    contrast to control samples, we identified a total of 199,372 differentially 17

    methylated CpG sites (CpGs) in FH-deficient RCCs, of which 66% (132,220) 18

    were hypermethylated CpG sites, whereas only 34% (67,152) were 19

    hypomethylated CpGs (Figure 2A, Figure S4B and C). 20

    Gene ontology enrichment analysis revealed that hypermethylated CpGs 21

    were significantly enriched in genes related to DNA-binding transcription 22

    activity and sequence-specific DNA binding (Figure 2B), indicating the 23

    transcriptional regulators might participate in the pathogenesis of FH-deficient 24

    RCC. Moreover, we found part of hypermethylated CpGs located in multiple 25

    well-established tumor suppressor genes (CDKN2A, APC, MGMT and TP53) 26

    and small RNAs (MIR200a, MIR200b, MIR200c and MIR429) (Figure S4D 27

    and E). Bisulfite pyrosequencing was used to validate DNA methylation levels 28

    in four gene regions in three hypermethylated loci (CDKN2A, APC and MGMT) 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 13

    and one hypomethylated locus (IL4R). A high correlation was observed 1

    between values obtained from microarray and pyrosequencing (Figure S4F). 2

    Furthermore, functional enrichment analysis identified an enrichment of genes 3

    of hallmark pathways in cancer for aberrant methylation, including WNT, 4

    mTOR, TGF-β, NOTCH and EMT pathways (Figure 2B and Figure S4G). 5

    Hypermethylated CpGs also mapped to genes involving the DNA-damage 6

    repair pathways (Figure S4H). Gene set enrichment analysis (GSEA) using 7

    genome array data further revealed that genes linked to EMT, mTORC1, 8

    DNA-damage repair and glycolysis pathways were upregulated in FH-mutated 9

    RCCs versus normal tissues (Figure 2C). 10

    11

    Identification of the methylation patterns of FH-deficient RCC 12

    To illuminate the DNA methylation pattern of FH-deficient RCC, we 13

    merged data from our FH-deficient RCC and TCGA-KIRP cohorts. Using 14

    unsupervised clustering analysis, we identified three DNA methylation clusters 15

    (Figure 2D). The majority of our FH-deficient RCC cases (80%,16/20) and all 16

    of the CIMP-RCC cases in KIRP cohort were categorized into the CIMP cluster, 17

    exhibiting a global DNA hypermethylation phenotype. This finding supports 18

    that most FH-deficient RCCs are largely characterized by a CIMP. However, a 19

    small proportion of our FH-deficient RCC cases (20%,4/20) were classified into 20

    cluster 1, manifested a relatively low genome-wide DNA methylation, indicating 21

    that not all FH-deficient RCCs exhibited CIMP. To further validated these 22

    findings, a T-distributed stochastic neighbor embedding analysis was 23

    performed to integrated ours and all TCGA RCC subtypes data. The results 24

    confirmed that the majority of FH-deficient RCCs were distributed in the 25

    CIMP-RCC cluster, while the presence of two different DNA methylation 26

    profiles existed in our FH-deficient RCCs (Figure 2E). Notably, we found 27

    patients in the non-CIMP group had a lower incidence of metastasis compared 28

    to the CIMP group (25% vs. 94%, P = 0.013). Three patients in the non-CIMP 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 14

    group did not develop recurrences or metastases up to the end of follow-up 1

    (median: 19.7 months), with one patient (FHRCC03) followed for over six 2

    years after nephrectomy. Morphologically, the majority of CIMP tumors 3

    (69%,11/16) showed a predominated papillary pattern, but only one non-CIMP 4

    tumor (25%, 1/4) showed a dominated papillary pattern. Two non-CIMP tumors 5

    (FHRCC03 and FHRCC12) were presented with a dominated low-grade 6

    oncocytic pattern, which has been reported with a favorable outcome (2,6,7). 7

    Methylation profiling analysis indicated that CIMP tumors had sharp global 8

    hypermethylation compared to non-CIMP tumors in all epigenomic structures, 9

    except for open seas (Figure S5A and B). Signature analysis further identified 10

    a total of 234,563 different signatures between these two methylation groups 11

    (Figure S5C). 12

    13

    FH-deficient RCC manifests high immune infiltration and imprints of 14

    immune evasion 15

    As mentioned above, a fraction of CpGs were hypomethylated in 16

    FH-deficient RCC. Strikingly, these hypomethylated CpGs were enriched in 17

    genes involving the inflammation pathway and cytokine activity (Figure 3A). 18

    GSEA analysis of the genome array further validated the increased expression 19

    of TNF-α-NF-κB and inflammatory response signaling in FH-mutated RCC, 20

    indicating the enhanced anti-tumor inflammatory signature in FH-deficient 21

    RCC (Figure 3A). Using immunohistochemistry (IHC) staining, we identified 22

    that the majority of tumors (96%, 24/25) showed enriched CD8+ T and CD4+ T 23

    cell infiltration. Sixty-four percent (16/25) of tumors had extensive CD8+ T cell 24

    infiltration (CD8+ T cells ≥ 100 mm2) (Table S4 and Figure 3B). In situ multiple 25

    immunofluorescence staining further revealed the increased immune-infiltrated 26

    in FH-deficient RCC, with marked activated NK (CD56+) cells and 27

    macrophages (CD68+/HLA-DR+) infiltrations in tumor stroma (Figure 3C). 28

    Importantly, GSEA analysis confirmed that IFN-γ response and IFN-γ 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 15

    dependent cancer immunostimulation pathways (IL-6–JAK-STAT3) were 1

    significantly enriched in FH-mutated RCC, indicating the presence of activated 2

    T cell signature. (Figure S6A). 3

    Considering that the production of IFN-γ can induce PD-L1 expression 4

    and subsequently leads to immune evasion (8), we next checked the 5

    expression of PD-L1 in FH-deficient RCC. Intriguingly, we found that PD-L1 6

    associated CpGs were hypomethylated modified in tumors (Figure S6B). In 7

    addition, hypomethylated CpGs also mapped to other immune inhibitory 8

    checkpoint genes (including PDCD1L2 and CTLA4, Figure 3D and Figure 9

    S6B). IHC experiments further demonstrated a high PD-L1 positive rate (80%, 10

    20/25) in tumors (Table S4). Of note, nearly half of tumors (48%,12/25) 11

    exhibited type II tumor immune microenvironment (TIME) (9), characterized by 12

    massive CD8+ T cell infiltration but positive PD-L1 expression in tumors 13

    (Figure 3D). Collectively, our data demonstrated that the anti-tumor immunity 14

    in FH-deficient RCC might be largely blocked by PD-1/PD-L1 pathway 15

    mediated suppression mechanism, suggesting the potential of PD-1/PD-L1 16

    inhibitors. 17

    18

    Patients with metastatic FH-deficient RCC benefit from immune 19

    checkpoint blockade-based immunotherapy 20

    At the end of follow-up, 18 patients with metastatic diseases received 21

    systemic therapy. First-line therapy was tyrosine kinase inhibitor (TKI) 22

    monotherapy (67%, 12/18), anti-PD-1 inhibitor plus TKI (27%, 5/18) and 23

    anti-PD-1 inhibitor monotherapy (6%, 1/18). For those receiving first-line 24

    therapy, the DCR was higher in patients receiving immune checkpoint 25

    blockade (ICB)-based treatment (anti-PD-1 inhibitor + TKIs or anti-PD-1 26

    inhibitor monotherapy) versus those on TKIs monotherapy (DCR: 100% vs. 27

    58.3%, Figure 4A). Moreover, median first-line PFS was superior in the 28

    ICB-based treatment group compared with that in TKIs group (median: 13.3 vs. 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 16

    5.1 months, P = 0.024, Figure 4B). All patients (6/6, 100%) in the ICB-based 1

    first-line treatment group achieved a PFS ≥ 6 months, higher than those (5/12, 2

    42%) in the TKIs group (Figure 4C and a typical case was illustrated in Figure 3

    4D). The median OS of systematic treatment was also longer in the ICB-based 4

    group compared to the TKIs group (median: not reached vs. 21.4 months); 5

    however, the differences did not reach a statistical significance (P = 0.209, 6

    Figure 4B). Two patients received additional anti-PD-1 therapy plus TKIs after 7

    disease progression during first-line TKIs monotherapy; remarkably, both of 8

    them achieved favorable disease control (PFS: 13.5 and 10.3 months, 9

    respectively). 10

    11

    Discussion 12

    FH-deficient RCC is a rare but lethal malignancy, and its genetic basis is 13

    poorly understood. In the present study, we performed comprehensive 14

    molecular profiling to characterize the genomic foundation of FH-deficient RCC. 15

    We determined that FH-deficient RCC was a genetically simple tumor with a 16

    relatively low mutation burden, but its distinct epigenetic features could be the 17

    biological foundation for tumor pathogenesis. Moreover, FH-deficient RCC was 18

    an intrinsically immunogenic tumor that is likely to benefit from ICB-based 19

    treatment strategies. Our study provides novel insights into the molecular 20

    landscape of FH-deficient RCC and identifies promising therapeutic targets for 21

    potential exploitation. 22

    Although with low somatic mutation burden, we identified increased 23

    SCNAs in FH-deficient RCC, with significant loss of 1p, 8q, 10p, 15q and gain 24

    of 4p, 7q, 11q, and 19p. Previous studies identified that a subgroup of papillary 25

    type 2 RCCs were characterized by a high degree of aneuploidy and frequent 26

    loss of 9p (4), which was not significantly altered in our FH-deficient RCCs. 27

    The different SCNAs patterns between FH-deficient RCC and papillary type 2 28

    RCC indicates the distinct genomic alterations during tumorigenesis, although 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 17

    the majority of FH-deficient RCC was predominantly of type 2 histology. 1

    Recent studies also showed an increased chromosome 22 loss in CIMP-RCC 2

    (10), however, it was not a significant event in our cohort. This discrepancy 3

    may reflect the heterogeneity of CIMP-RCC and warrant further investigation. 4

    Loss-of-function mutations in FH resulting in the accumulation of fumarate 5

    impairs the function of α-KG-dependent dioxygenases, such as histone and 6

    DNA demethylases, which disrupts the normal epigenetic genes regulation 7

    (11). Consistent with this, we identified FH-deficient RCC-specific DNA 8

    methylation changes within a broad panel of transcription factors, as well as 9

    genes significantly associated with tumor progression and patient outcomes, 10

    including CDKN2A, MGMT, APC and TP53. The loss of CDKN2A by either 11

    mutation, deletion or promoter hypermethylation occurred in 15.8% RCCs and 12

    correlated with decreased survival in all RCC histological subtypes (10). Of 13

    note, we observed that CDKN2A promoter hypermethylation is a common 14

    event in FH-deficient RCCs. Loss of CDKN2A can be targeted by inhibitors of 15

    cyclin-dependent kinase 4/6 (CDK4/6) (12), suggesting the potential of 16

    CDK4/6 inhibitors for FH-deficient RCCs. Also, it should be pointed out that it 17

    remains unclear if the loss of CDKN2A could be a predictor for response to 18

    CDK4/6 inhibitors, and still warrant further investigation(13). Currently, 19

    epigenetic agents have been approved for the treatment of hematological 20

    malignancies (14) and are now undergoing testing in HLRCC 21

    (ClinicalTrials.gov identifier NCT03165721). Our data imply that these aberrant 22

    DNA methylations could be the biological foundation of FH-deficient RCC, 23

    highlighting the potential of demethylation agents for treating this disease. 24

    Another important finding of our study is that FH-deficient RCC exhibits a 25

    distinct immunophenotype. Albeit with low mutation burden, most FH-deficient 26

    RCCs exhibited typical types II TIME, characterized by increased levels of 27

    tumor infiltrating lymphocytes and activated chemotaxis signaling, but with 28

    high expression level of PD-L1. Therefore, it is reasonable to assume that 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 18

    anti-PD-1/PD-L1 therapy can be a potential treatment option for patients with 1

    FH-deficient RCC (9,15). Indeed, we observed a favorable PFS for patients 2

    receiving ICB-based treatment compared to those receiving antiangiogenic 3

    monotherapy. Mechanistically, this immunogenicity may be mediated by 4

    epigenomic and metabolic alterations. Using the TCGA data, Singh et al. found 5

    that hypomethylation of genes was associated with immune function in 6

    papillary RCC (16). Recent studies also show that tricarboxylic acid cycle 7

    metabolites, including fumarate, can regulate methylation status in immune 8

    cells and facilitate transcription and expression of proinflammatory factors 9

    (17-20). Similarly, we observed epigenetically enhanced genes involved in 10

    pro-inflammatory and immune suppression pathways in FH-deficient RCC. 11

    Taken together, our data suggest that FH-deficient RCC is intrinsically 12

    immunogenic and could be targeted by ICB-based immunotherapy. Given the 13

    global hypermethylation phenotype, co-administration of epigenomic agents 14

    could be a promising strategy to improve the efficacy of PD-1/PD-L1 inhibitors. 15

    In addition to the treatments mentioned above, there are other potential 16

    therapeutic targets according to our findings. Mutation of the Hippo pathway 17

    tumor suppressor, NF2, was detected in several tumors. Thus, targeting NF2 18

    (e.g. Dasatinib) (21) may be effective in a subset of patients. Moreover, 19

    preclinical studies suggested that elevated fumarate could suppress the 20

    homologous recombination DNA repair pathway through inhibition of 21

    demethylases (22,23). We observed mutation and frequently aberrant 22

    methylation of genes involved in DNA damage repair pathways in FH-deficient 23

    RCC, which supports the poly (ADP)-ribose polymerase inhibitors for targeted 24

    therapy. Additionally, both ours and previous studies showed the activation of 25

    glycolysis (4,24,25) and mTOR signaling in FH-deficient RCCs. Results from a 26

    single-center phase II trial (ClinicalTrials.gov Identifier: NCT01130519) and 27

    retrospective studies (26,27) reported the combination of bevacizumab plus 28

    erlotinib could achieve fair objective response for patients with FH-deficient 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 19

    RCC, indicating the potential therapeutic value of this combination therapy. 1

    In summary, the current study expands our knowledge of the FH-deficient 2

    RCC somatic alteration landscape, emphasizes the importance of DNA 3

    methylation profiles for tumor pathogenesis and molecular classification, and 4

    reveals its immunogenic phenotype. Our study represents a step forward in 5

    understanding the biology of FH-deficient RCC and provides a genetic basis 6

    for biomarker classification and potential therapeutic strategies. 7

    8

    Acknowledgements 9

    This work was supported by the Natural Science Foundation of China (NSFC 10

    81672547, 81872107, 81872108, 81974398, 81972502 and 81902577), China 11

    Postdoctoral Science Foundation (2020M673239) and 1.3.5 project for 12

    disciplines of excellence, West China Hospital, Sichuan University 13

    (No.0040205301E21). The authors thank all patients involved in this study, as 14

    well as their families/caregivers. We offer sincere gratitude to Prof. Allen C. 15

    Gao from UC Davis Medical Center for the constructive suggestions and 16

    careful revision for this manuscript. 17

    18

    Author contributions 19

    Study concept and design: S.G.X., X.M.Z., J.Y.L., X.Y.P., S.Z., Z.H.L., H.J.H, 20

    J.Y., N.C., H.Z.; 21

    Methodology, acquisition, analysis, or interpretation of the data: S.G.X., X.M.Z., 22

    J.Y.L., X.Y.P., S.Z., Z.H.L., J.H.C., W.L., B.H.L., T.H.L., R.H., X.X.Y., M.N.Z., 23

    L.N., P.F.S., J.G.Z., H.R.Z., J.D.D., Y.L.S., Z.P.L., J.Y.L., J.R.C., J.D.L., Z.P.W., 24

    X.D.Z., Y.C.N., D.Q., L.Y., Y.T.C., H.J.H, J.Y., N.C., H.Z.; 25

    Financial support: S.G.X., R.H., Q.Z., N.C., H.Z.; 26

    Drafting of the manuscript: S.G.X., X.M.Z., J.Y.L., X.Y.P., S.Z., Z.H.L.; 27

    Critical revision of the manuscript for important intellectual content: S.G.X., 28

    X.M.Z., J.Y.L., X.Y.P., S.Z., Z.H.L., H.J.H, C.M. A., Z.H.L., J.Y., N.C., H.Z.; 29

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 20

    Statistical analysis: S.G.X., X.M.Z., J.Y.L., X.Y.P., S.Z., Z.H.L., J.G.Z. 1

    Study supervision: Q.W, X.L., Q.Z., H.J.H, J.Y., N.C., H.Z. 2

    3

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 21

    Reference:

    1. Trpkov K, Hes O, Agaimy A, Bonert M, Martinek P, Magi-Galluzzi C, et

    al. Fumarate Hydratase-deficient Renal Cell Carcinoma Is Strongly

    Correlated With Fumarate Hydratase Mutation and Hereditary

    Leiomyomatosis and Renal Cell Carcinoma Syndrome. Am J Surg

    Pathol 2016;40(7):865-75 doi 10.1097/pas.0000000000000617.

    2. Lau HD, Chan E, Fan AC, Kunder CA, Williamson SR, Zhou M, et al. A

    Clinicopathologic and Molecular Analysis of Fumarate

    Hydratase-deficient Renal Cell Carcinoma in 32 Patients. Am J Surg

    Pathol 2020;44(1):98-110 doi 10.1097/PAS.0000000000001372.

    3. Grubb RL, 3rd, Franks ME, Toro J, Middelton L, Choyke L, Fowler S, et

    al. Hereditary leiomyomatosis and renal cell cancer: a syndrome

    associated with an aggressive form of inherited renal cancer. J Urol

    2007;177(6):2074-9; discussion 9-80 doi 10.1016/j.juro.2007.01.155.

    4. Cancer Genome Atlas Research N, Linehan WM, Spellman PT, Ricketts

    CJ, Creighton CJ, Fei SS, et al. Comprehensive Molecular

    Characterization of Papillary Renal-Cell Carcinoma. N Engl J Med

    2016;374(2):135-45 doi 10.1056/NEJMoa1505917.

    5. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford

    R, et al. New response evaluation criteria in solid tumours: revised

    RECIST guideline (version 1.1). Eur J Cancer 2009;45(2):228-47 doi

    10.1016/j.ejca.2008.10.026.

    6. Smith SC, Sirohi D, Ohe C, McHugh JB, Hornick JL, Kalariya J, et al. A

    distinctive, low-grade oncocytic fumarate hydratase-deficient renal cell

    carcinoma, morphologically reminiscent of succinate

    dehydrogenase-deficient renal cell carcinoma. Histopathology

    2017;71(1):42-52 doi 10.1111/his.13183.

    7. Li Y, Reuter VE, Matoso A, Netto GJ, Epstein JI, Argani P. Re-evaluation

    of 33 'unclassified' eosinophilic renal cell carcinomas in young patients.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 22

    Histopathology 2018;72(4):588-600 doi 10.1111/his.13395.

    8. Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response

    to immune checkpoint blockade. Nat Med 2019;25(3):389-402 doi

    10.1038/s41591-019-0382-x.

    9. Sanmamed MF, Chen L. A Paradigm Shift in Cancer Immunotherapy:

    From Enhancement to Normalization. Cell 2018;175(2):313-26 doi

    10.1016/j.cell.2018.09.035.

    10. Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, et al.

    The Cancer Genome Atlas Comprehensive Molecular Characterization

    of Renal Cell Carcinoma. Cell Rep 2018;23(1):313-26 e5 doi

    10.1016/j.celrep.2018.03.075.

    11. Xiao M, Yang H, Xu W, Ma S, Lin H, Zhu H, et al. Inhibition of

    alpha-KG-dependent histone and DNA demethylases by fumarate and

    succinate that are accumulated in mutations of FH and SDH tumor

    suppressors. Genes Dev 2012;26(12):1326-38 doi

    10.1101/gad.191056.112.

    12. Rugo HS, Rumble RB, Macrae E, Barton DL, Connolly HK, Dickler MN,

    et al. Endocrine Therapy for Hormone Receptor-Positive Metastatic

    Breast Cancer: American Society of Clinical Oncology Guideline. J Clin

    Oncol 2016;34(25):3069-103 doi 10.1200/JCO.2016.67.1487.

    13. Baghdadi TA, Halabi S, Garrett-Mayer E, Mangat PK, Ahn ER, Sahai V,

    et al. Palbociclib in Patients With Pancreatic and Biliary Cancer With

    CDKN2A Alterations: Results From the Targeted Agent and Profiling

    Utilization Registry Study. JCO Precision Oncology 2019(3):1-8 doi

    10.1200/po.19.00124.

    14. Fenaux P, Mufti GJ, Hellström-Lindberg E, Santini V, Gattermann N,

    Germing U, et al. Azacitidine Prolongs Overall Survival Compared With

    Conventional Care Regimens in Elderly Patients With Low Bone

    Marrow Blast Count Acute Myeloid Leukemia. Journal of Clinical

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 23

    Oncology 2010;28(4):562-9 doi 10.1200/jco.2009.23.8329.

    15. Doroshow DB, Sanmamed MF, Hastings K, Politi K, Rimm DL, Chen L,

    et al. Immunotherapy in Non-Small Cell Lung Cancer: Facts and Hopes.

    Clin Cancer Res 2019;25(15):4592-602 doi

    10.1158/1078-0432.CCR-18-1538.

    16. Singh NP, Vinod PK. Integrative analysis of DNA methylation and gene

    expression in papillary renal cell carcinoma. Mol Genet Genomics

    2020;295(3):807-24 doi 10.1007/s00438-020-01664-y.

    17. Arts RJW, Novakovic B, ter Horst R, Carvalho A, Bekkering S,

    Lachmandas E, et al. Glutaminolysis and Fumarate Accumulation

    Integrate Immunometabolic and Epigenetic Programs in Trained

    Immunity. Cell Metabolism 2016;24(6):807-19 doi

    10.1016/j.cmet.2016.10.008.

    18. Cordes T, Wallace M, Michelucci A, Divakaruni AS, Sapcariu SC, Sousa

    C, et al. Immunoresponsive Gene 1 and Itaconate Inhibit Succinate

    Dehydrogenase to Modulate Intracellular Succinate Levels. J Biol Chem

    2016;291(27):14274-84 doi 10.1074/jbc.M115.685792.

    19. Lampropoulou V, Sergushichev A, Bambouskova M, Nair S, Vincent

    Emma E, Loginicheva E, et al. Itaconate Links Inhibition of Succinate

    Dehydrogenase with Macrophage Metabolic Remodeling and

    Regulation of Inflammation. Cell Metabolism 2016;24(1):158-66 doi

    10.1016/j.cmet.2016.06.004.

    20. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M,

    Fuchs E, et al. Defining trained immunity and its role in health and

    disease. Nature Reviews Immunology 2020 doi

    10.1038/s41577-020-0285-6.

    21. R J, G H. The two faces of Hippo: targeting the Hippo pathway for

    regenerative medicine and cancer treatment. Nature reviews Drug

    discovery 2014;13(1):63-79 doi 10.1038/nrd4161.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 24

    22. Sulkowski PL, Sundaram RK, Oeck S, Corso CD, Liu Y, Noorbakhsh S,

    et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by

    defects in homologous-recombination DNA repair. Nat Genet

    2018;50(8):1086-92 doi 10.1038/s41588-018-0170-4.

    23. Sulkowski PL, Oeck S, Dow J, Economos NG, Mirfakhraie L, Liu Y, et al.

    Oncometabolites suppress DNA repair by disrupting local chromatin

    signalling. Nature 2020;582(7813):586-91 doi

    10.1038/s41586-020-2363-0.

    24. S S, C S, HS K, K B, VA VR, Y Y, et al. Fumarate hydratase deficiency

    in renal cancer induces glycolytic addiction and hypoxia-inducible

    transcription factor 1alpha stabilization by glucose-dependent

    generation of reactive oxygen species. Molecular and cellular biology

    2009;29(15):4080-90 doi 10.1128/mcb.00483-09.

    25. WH T, C S, G K, SY J, M V, M G, et al. The glycolytic shift in

    fumarate-hydratase-deficient kidney cancer lowers AMPK levels,

    increases anabolic propensities and lowers cellular iron levels. Cancer

    cell 2011;20(3):315-27 doi 10.1016/j.ccr.2011.07.018.

    26. Choi Y, Keam B, Kim M, Yoon S, Kim D, Choi JG, et al. Bevacizumab

    Plus Erlotinib Combination Therapy for Advanced Hereditary

    Leiomyomatosis and Renal Cell Carcinoma-Associated Renal Cell

    Carcinoma: A Multicenter Retrospective Analysis in Korean Patients.

    Cancer Research and Treatment 2019;51(4):1549-56 doi

    10.4143/crt.2019.086.

    27. Park I, Shim YS, Go H, Hong BS, Lee JL. Long-term response of

    metastatic hereditary leiomyomatosis and renal cell carcinoma

    syndrome associated renal cell carcinoma to bevacizumab plus erlotinib

    after temsirolimus and axitinib treatment failures. BMC urology

    2019;19(1):51 doi 10.1186/s12894-019-0484-2.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 25

    Figure legends

    Figure 1. Somatic alterations in FH-deficient RCC.

    (A) Oncoplot describes somatic genomic alterations of the FH-deficient RCC

    (n = 24). Recurrently mutated genes, which are grouped by molecular

    pathways, are shown on the left. Their mutation frequencies in patients with

    onset of RCC < 40 years and ≥ 40 years old are shown on the right. The top

    histogram shows the number of somatic mutations in each individual sample.

    The middle panel shows the clinicopathological features. The bottom

    histogram shows the mutation spectrum. Red arrow indicates two samples

    (FHRCC18 and FHRCC24) performing panel test. See also Table S1 and S4.

    (B) The types and relative positions of somatic mutations in the predicted FH

    protein.

    (C) Copy-number profile shows amplifications (red) and deletions (blue) in all

    chromosomes for FH-deficient RCC. Cancer-related genes located within

    corresponding genomic regions are indicated. For genes located in 19

    chromosome, the detailed locations are: AKT2 (19q13.2), ARID3A (19p13.3),

    CDKN2D (19p13.2), ERCC1 (19q13.32), ERCC2 (19q13.32), KDM4B

    (19p13.3), SMARCA4 (19p13.2), XRCC1 (19q13.31).

    FH = fumarate hydratase, RCC = renal cell carcinoma, IHC =

    immunohistochemistry.

    Figure 2. FH-deficient RCCs manifest a distinct methylation phenotype.

    (A) A volcano plot comparing DNA methylation in tumors versus paired

    adjacent normal tissues. Significantly hypermethylated CpG sites are colored

    in red, whereas significantly hypomethylated CpG sites are colored in blue. q

    values (-log10 p) were calculated using paired two-sided moderated Student’s

    t-test.

    (B) Gene ontology enrichment analysis of differentially methylated CpG sites.

    Enrichment q values (-log10 p) are calculated by hypergeometric test. See also

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 26

    Figure S4.

    (C) GSEA enrichment scores of glycolysis, EMT, DNA-damage repair and

    mTORC1 pathways in HLRCC versus normal tissues.

    (D) One-dimensional hierarchical clustering of the 2,000 most variant DNA

    methylation probes reveal three clusters of tumors from TCGA KIRP cohort (n

    = 275) and our FH-deficient RCC cohort (n = 20). In contrast to cluster 1 and 2,

    cluster 3 shows widespread DNA hypermethylation patterns characteristic of

    CIMP-associated tumors. Adjacent normal tissues (n = 12) are included in the

    heatmap but did not contribute to the unsupervised clustering. Each row

    represents a probe; each column represents a sample.

    (E) T-distributed stochastic neighbor embedding analysis of 842 TCGA RCC

    samples and 20 FH-deficient RCC samples performed on 2,000 reference

    probes illustrates three distinct clusters.

    FH = fumarate hydratase, RCC = renal cell carcinoma, GSEA = gene set

    enrichment analyses, EMT = epithelial-mesenchymal transition, TCGA = The

    Cancer Genome Atlas, HLRCC = hereditary leiomyomatosis and renal cell

    carcinoma, KICH = chromophobe renal cell carcinoma, KIRC = renal clear cell

    carcinoma, KIRP = renal papillary cell carcinoma, CIMP = CpG island

    methylator phenotype.

    Figure 3. FH-deficient RCCs exhibit intrinsic immunogenic phenotype

    (A) Bubble chart (upper) shows the significantly hypomethylated CpGs in

    FH-deficient RCCs. Enrichment q values (-log10 p) are calculated by

    hypergeometric test. GSEA enrichment scores (lower) of TNF-α-NF-κB and

    inflammation response signaling in HLRCC versus normal tissues. See also

    Figure S6.

    (B) Representative IHC demonstrating the typical TIME type II in CIMP

    (FHRCC13) and non-CIMP tumors (FHRCC12) in our cohort, is characterized

    by PD-L1 positive expression and massive CD8+ and CD4+T cell infiltrations.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 27

    TIME type II is defined as PD-L1 positive in tumors and massive CD8+ T cell

    infiltration (>100 cells/mm2). Magnification X200. Scale bar = 100μm.

    (C) Representative immunofluorescence demonstrating the presence of

    overall CD8+ T cell, tumor-associated macrophage (CD68 and HLA-DR) and

    NK cell (CD56) infiltration in two selected samples (FHRCC16 and FHRCC19)

    in our cohort. Magnification X400. Scale bar = 100μm.

    (D) Heatmap shows the immune features among FH-deficient RCCs (n = 25).

    The top panel shows the number of CD8+ and CD4+ T cell infiltrations, PD-L1

    expression and TIME type. The middle panel shows the methylation status of a

    CpG site in the promoter region of immune modulation associated genes. The

    bottom histogram shows the FH mutation type, FH protein expression and

    CIMP status.

    FH = fumarate hydratase, RCC = renal cell carcinoma, HLRCC = hereditary

    leiomyomatosis and renal cell carcinoma, GSEA = gene set enrichment

    analyses, TIME = tumor immune microenvironment, CIMP = CpG island

    methylator phenotype.

    Figure 4. Responses to systemic treatment in patients with metastatic

    FH-deficient RCC.

    (A) Histogram shows the best radiographic response in patients receiving

    first-line TKIs and ICB-based treatment groups. The best response is

    assessed via Response Evaluation Criteria in Solid Tumors 1.1. ICB-based

    treatments include patients receiving anti-PD-1 inhibitor plus TKIs or anti-PD-1

    inhibitor monotherapy.

    (B) Kaplan-Meier curves show the first-line PFS (left) and OS (right) between

    patients receiving first-line TKIs (red) and ICB-based (green) treatments.

    (C) Swimmer plot depicts the PFS of individual patients receiving first-line

    systematic treatments. Each patient’s tumor response at the end of follow-up

    (PR, SD or PD) is noted. Vertical line indicates PFS at 6 months.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • 28

    (D) Baseline imaging in a patient (FHRCC16) before initiation of systematic

    treatment, with multiple para-aorta lymphatic metastases. The patient

    subsequently received the combination of Sintilimab plus Axitinib treatment.

    Imaging performed at 3 months after the combination therapy demonstrates

    continued objective response (33.9% by RECIST v1.1).

    FH = fumarate hydratase, RCC = renal cell carcinoma, PR = partial response,

    SD = stable disease, PD = progression disease, ORR = objective response

    rate, PFS = progression-free survival, OS = overall survival, TKIs = tyrosine

    kinase inhibitors, ICB = immune checkpoint blockade.

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/

  • Table 1. Baseline characteristics of patients with FH-deficient RCC

    Patient Characteristics N(%)

    Total number of patients 25(100)

    Age at diagnosis, years (Median, range) 38(13-66)

    Male 16(64)

    Female 9(36)

    Patients with skin leiomyoma 2(8)

    Female patients with uterine leiomyoma 5(55.6)

    Primary tumor size, cm (Median, range) 8.8(3-16.9)

    T stage

  • Published OnlineFirst January 7, 2021.Clin Cancer Res Guangxi Sun, Xingming Zhang, Jiayu Liang, et al. Hydratase-deficient Renal Cell CarcinomaIntegrated Molecular Characterization of Fumarate

    Updated version

    10.1158/1078-0432.CCR-20-3788doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2021/01/07/1078-0432.CCR-20-3788.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authorbeen edited. Author manuscripts have been peer reviewed and accepted for publication but have not yet

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2021/01/07/1078-0432.CCR-20-3788To request permission to re-use all or part of this article, use this link

    Research. on July 3, 2021. © 2021 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on January 7, 2021; DOI: 10.1158/1078-0432.CCR-20-3788

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-20-3788http://clincancerres.aacrjournals.org/content/suppl/2021/01/07/1078-0432.CCR-20-3788.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2021/01/07/1078-0432.CCR-20-3788http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Table 1